US20210355229A1 - An anti-ox40 antibody, antigen-binding fragment thereof, and the pharmaceutical use - Google Patents

An anti-ox40 antibody, antigen-binding fragment thereof, and the pharmaceutical use Download PDF

Info

Publication number
US20210355229A1
US20210355229A1 US17/278,259 US201917278259A US2021355229A1 US 20210355229 A1 US20210355229 A1 US 20210355229A1 US 201917278259 A US201917278259 A US 201917278259A US 2021355229 A1 US2021355229 A1 US 2021355229A1
Authority
US
United States
Prior art keywords
seq
antibody
antigen
binding fragment
variable region
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/278,259
Other languages
English (en)
Inventor
Cheng Liao
Zupeng Xu
Jiahua JIANG
Xin Ye
Lianshan Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jiangsu Hengrui Medicine Co Ltd
Original Assignee
Jiangsu Hengrui Medicine Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jiangsu Hengrui Medicine Co Ltd filed Critical Jiangsu Hengrui Medicine Co Ltd
Assigned to JIANGSU HENGRUI MEDICINE CO., LTD. reassignment JIANGSU HENGRUI MEDICINE CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JIANG, Jiahua, LIAO, Cheng, XU, Zupeng, YE, XIN, ZHANG, LIANSHAN
Publication of US20210355229A1 publication Critical patent/US20210355229A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/80Vectors or expression systems specially adapted for eukaryotic hosts for fungi
    • C12N15/81Vectors or expression systems specially adapted for eukaryotic hosts for fungi for yeasts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57492Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds localized on the membrane of tumor or cancer cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/577Immunoassay; Biospecific binding assay; Materials therefor involving monoclonal antibodies binding reaction mechanisms characterised by the use of monoclonal antibodies; monoclonal antibodies per se are classified with their corresponding antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • the present invention belongs to the field of biomedicine and relates to an anti-OX40 antibody, antigen-binding fragment thereof, a chimeric antibody, a humanized antibody comprising the CDR regions of the anti-OX40 antibody, and a pharmaceutical composition comprising the anti-OX40 antibody and antigen-binding fragment thereof, and the use of the same as an anti-cancer agent.
  • Cancer is a severe health challenge faced by human society for a long time.
  • traditional surgery, chemotherapy, and radiotherapy usually exhibit limited effect.
  • Tumor immunotherapy is continuously a hot spot in the field of tumor therapy. Recent studies have proved that enhancing the function of anti-tumor T cells can be used to counteract cancer. There is a lot of evidence showing that tumor cells “escape” from the immune system by inducing active immune tolerance mediated mainly by regulatory T lymphocytes (Treg; Quezda et al. Immunol Rev 2011; 241:104-118). Therefore, the balance between effector T lymphocytes (Teff) and tolerogenic Treg is essential for effective anti-tumor immunotherapy. Therefore, an effective anti-tumor immune response can be obtained by enhancing the effector function of tumor-specific Teff and/or by reducing the inhibitory function of tumor-specific Treg.
  • Treg regulatory T lymphocytes
  • the CD134 (OX40) receptor has been shown to be a key receptor that mediates these responses (Sugamura, K, Ishii, N, Weinberg, A. Therapeutic targeting of the effector T-cell co-stimulatory molecule OX40. Nature Rev Imm 2004; 4:420-431).
  • OX40 is a member of the tumor necrosis factor receptor (TNFR) superfamily, and is a glycoprotein with a molecular weight of about 50kDa expressed on the cell surface.
  • the extracellular ligand-binding domain of OX40 consists of 4 cysteine-rich domains (CRD).
  • the natural ligand of OX40 is OX4OL (CD252), OX40 and OX4OL form an OX40-0X4OL complex.
  • OX40 is mainly expressed on activated T cells, and OX40 is a secondary costimulatory molecule that is expressed 24 to 72 hours after activation.
  • OX4OL a ligand of OX40, is mainly expressed on activated antigen-presenting cells.
  • T lymphocytes expressing OX40 have been confirmed to exist in the draining lymph nodes of patients with various human malignant tumors and cancers.
  • SCID severe combined immunodeficiency
  • the interaction of OX40 and OX4OL binding domains can enhance anti-tumor immunity, resulting in tumor growth inhibition of various human malignant tumor cell lines, such as lymphoma, prostate cancer, colon cancer and breast cancer.
  • OX40 antibodies activate immunity through specific stimulation, improve the patient's own immune system response to tumors, and achieve the purpose of killing tumor cells.
  • Related patents are such as W02013038191, WO2015153513, WO2016179517, WO2017096182, CN110078825A, WO2016196228 and so on. So far, the anti-OX40 antibodies developed by companies such as AstraZeneca and BMS have been in phase II clinical trials, and related products from companies such as Genentech and GSK were also in clinical trials.
  • the present disclosure provides an anti-OX40 antibody or antigen-binding fragment thereof, which specifically binds to human OX40, and comprises the CDRs shown below:
  • heavy chain HCDR1, HCDR2 and HCDR3 as shown in SEQ ID NO: 11, 34 and 13 respectively
  • light chain LCDR1, LCDR2 and LCDR3 as shown in SEQ ID NO: 14, 15 and 16 respectively.
  • the anti-OX40 antibody or antigen-binding fragment thereof is a murine antibody, chimeric antibody, humanized antibody, human antibody or fragment thereof; in particular, a humanized antibody.
  • the CDRs (comprising 3 heavy chain CDRs and 3 light chain CDRs) of the monoclonal antibody or antigen-binding fragment are allowed to comprise 3, 2 or 1 amino acid difference(s) (i.e., CDR variants).
  • the CDR variants are obtained by affinity maturation method.
  • the affinity (KD) of the monoclonal antibody or antigen-binding fragment to OX40 is less than 10 ⁇ 8 M, less than 10 ⁇ 9 M, or less than 10 1 ° M.
  • the murine antibody comprises a heavy chain variable region and a light chain variable region as shown below:
  • the humanized antibody comprises FR region(s) derived from human germline or mutant sequence(s) thereof.
  • the humanized antibody comprises any one of:
  • the anti-OX40 antibody or antigen-binding fragment thereof comprises:
  • the anti-OX40 antibody or antigen-binding fragment thereof comprises:
  • the anti-OX40 antibody or antigen-binding fragment thereof comprises variable regions as shown below:
  • the anti-OX40 antibody or antigen-binding fragment thereof comprises variable regions as shown below:
  • the anti-OX40 antibody comprises a constant region; in some particular embodiments, the antibody is a chimeric antibody or a humanized antibody, and the heavy chain constant region of the antibody is derived from human IgG1, IgG2, IgG3, or IgG4 or the mutant sequence(s) thereof, the light chain constant region is derived from human kappa, lambda chain or the mutant sequence(s) thereof; in other particular embodiments, the amino acid sequence of the heavy chain constant region is as shown in SEQ ID NO: 35 or has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO: 35; the amino acid sequence of the light chain constant region is as shown in SEQ ID NO: 36 or has at least 70%, at least 75%, at least 80%, at least 85%, at
  • the heavy chain amino acid sequence of the anti-OX40 antibody is as shown in SEQ ID NO: 39 or 37 or has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO: 39 or 37, and/or, the light chain amino acid sequence of the anti-OX40 is as shown in SEQ ID NO: 40 or 38 or has at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% sequence identity to SEQ ID NO: 40 or 38.
  • the anti-OX40 antibody comprises:
  • the present disclosure also provides an anti-OX40 antibody or antigen-binding fragment thereof, the anti-OX40 antibody or antigen-binding fragment thereof competes with any antibody or antigen-binding fragment thereof described above for the binding to human OX40, or competes with any antibody or antigen-binding fragment thereof described above for binding to the same OX40 epitope.
  • composition which comprises:
  • the pharmaceutical composition may be prepared in a unit dose form, and the unit dose may comprise 0.01 to 99% by weight of the anti-OX40 antibody or antigen-binding fragment thereof; or the amount of the monoclonal antibody or antigen-binding fragment thereof comprised in the pharmaceutical composition is from 0.1 to 2000 mg/unit dose. In some particular embodiments, the amount of the monoclonal antibody or antigen-binding fragment thereof is from 1 to 1000 mg.
  • the monoclonal antibody or antigen-binding fragment thereof may be the one and only active ingredient comprised in the composition; in other particular embodiments, the monoclonal antibody or antigen-binding fragment thereof is used in combination with other active ingredients.
  • the present disclosure also provides an isolated nucleic acid molecule, which encodes the anti-OX40 antibody or antigen-binding fragment thereof described above.
  • the complementary sequence of the nucleic acid molecule is also included in the scope of this application.
  • the present disclosure also provides a vector comprising the nucleic acid molecule described above.
  • the vector can be a eukaryotic expression vector, a prokaryotic expression vector or a viral vector.
  • the present disclosure also provides a host cell transformed with the vector described above, and the host cell is selected from the group consisting of prokaryotic cells and eukaryotic cells.
  • the host cell is a eukaryotic cell.
  • the host cell is a mammalian cell, wherein the mammalian cell comprises but not limited to CHO, HEK293, and NSO. It should be understood that the host cell of the present disclosure does not involve any cell capable of developing into humans.
  • the present disclosure also provides a method for preparing the anti-OX40 antibody or antigen-binding fragment thereof described above, the method comprises:
  • the present disclosure also provides a method for detecting or measuring human OX40, the method comprises:
  • the present disclosure also provides a reagent for detecting or measuring human OX40, the reagent comprises any one of the anti-OX40 antibodies or antigen-binding fragment thereof described above.
  • the present disclosure also provides a diagnostic agent for diseases related to human OX40, the diagnostic agent comprises the anti-OX40 antibody or antigen-binding fragment thereof described above.
  • the present disclosure also provides a method for diagnosing diseases related to human OX40, the method comprises detecting or measuring human OX40 or OX40-positive cells using the anti-OX40 antibody or antigen-binding fragment thereof described above.
  • the present disclosure also provides the use of the anti-OX40 antibody or antigen-binding fragment thereof in the preparation of a diagnostic agent for diseases related to human OX40.
  • the present disclosure also provides a method for treating disease or disorder, which comprises administering to a subject an effective amount of the anti-OX40 antibody or antigen-binding fragment thereof, or a pharmaceutical composition comprising the OX40 antibody or fragment thereof described above.
  • the disease or disorder may be cancer or cell-proliferative disease.
  • the cancer is lung cancer, prostate cancer, breast cancer, head and neck cancer, esophageal cancer, gastric carcinoma, colon cancer, colorectal cancer, bladder cancer, cervical cancer, uterine cancer, ovarian cancer, liver cancer, melanoma, kidney cancer, squamous cell carcinoma, hematological cancer or any other disease or disorder characterized by uncontrolled cell growth.
  • the hematological cancer comprises but is not limited to acute and chronic myelogenous leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, bone marrow tissue proliferative disease, multiple myeloma, Hodgkin's disease, non-Hodgkin's lymphoma, B-cell lymphoma, T-cell lymphoma, follicular center cell lymphoma and chronic myeloid leukemia.
  • the present disclosure also provides the use of the anti-OX40 antibody or antigen-binding fragment thereof described above, or the pharmaceutical composition comprising the OX40 antibody or fragment thereof described above, in the preparation of a medicine for enhancing the immune response in a human subject.
  • the present disclosure also provides the use of the anti-OX40 antibody or antigen-binding fragment thereof described above, or the pharmaceutical composition comprising the OX40 antibody or fragment thereof described above, in the preparation of a medicament for the treatment/prevention of cancer.
  • the present disclosure also provides the use of the anti-OX40 antibody or antigen-binding fragment thereof described above, or the pharmaceutical composition comprising the OX40 antibody or fragment thereof described above, in the preparation of a kit.
  • the enhanced immune response comprises an increase in the immunostimulatory/effector function of T effector cells, and/or a down-regulation of the immunosuppressive function of T regulatory cells.
  • the increase may be the result of cell proliferation
  • the down-regulation may be the result of absence of increase in cell number (or decrease in cell number).
  • the present disclosure provides the anti-human OX40 antibody or fragment thereof for use in one or more of the following: inhibiting Treg function (for example, inhibiting the suppressive function of Treg), killing OX40-expressing cells (for example, cells expressing high level of OX40), improving effector T cell function and/or improving memory T cell function, reducing tumor immunity, enhancing T cell function and/or reducing OX40-expressing cells.
  • Treg function for example, inhibiting the suppressive function of Treg
  • OX40-expressing cells for example, cells expressing high level of OX40
  • improving effector T cell function and/or improving memory T cell function for example, reducing tumor immunity, enhancing T cell function and/or reducing OX40-expressing cells.
  • the present disclosure provides the use of the anti-OX40 antibody or fragment thereof for the following: treating cancer, stimulating immune response in a subject, stimulating antigen-specific T cell response, activating or co-stimulating T cells, increasing cytokines production in T cells (such as IL-2 and/or IFN- ⁇ ), and/or increasing T cell proliferation, reducing or depleting the number of T regulatory cells in tumor, and/or inhibiting the growth of tumor cells.
  • the present disclosure also provides the use of the anti-OX40 antibody in the preparation of an agent, the agent is for use in stimulating immune response in a subject, stimulating antigen-specific T cell response, activating or co-stimulating T cells, increasing IL-2 and/or IFN- ⁇ production in T cells and/or T cell proliferation, reducing or depleting the number of T regulatory cells in tumor, and/or inhibiting the growth of tumor cells.
  • FIG. 1A to FIG. 1B ELISA test results showing the affinity of murine antibody and chimeric antibody to human OX40.
  • FIG. 1A shows the result of the affinity of the murine antibody m2G3 and the chimeric antibody ch2G3, with m2G3-NC and ch2G3-NC as negative controls;
  • FIG. 1B shows the result of the affinity of the murine antibody m4B5 and the chimeric antibody ch4B5, with m4B5-NC and ch4B5-NC as negative controls.
  • FIG. 2 Test showing the IFN- ⁇ secreted in T cells stimulated by anti-OX40 antibody. The results show that the test OX40 antibody achieves the maximum stimulating effect at a concentration of 10 ng/mL.
  • FIG. 3A to FIG. 3B anti-tumor effect of anti-OX40 antibody in mice.
  • FIG. 3A shows the changes in tumor volume in different mice after administration;
  • FIG. 3B shows the effect of different anti-OX40 antibodies on tumor weight in mice after administration. The results show that on day 20 after dosing treatment, when the dose is at 3 mg/kg, the tumor inhibition rate of 2G3 antibody is up to 97%.
  • enhancing T cell function means inducing, causing or stimulating effector or memory T cells, so that they have renewal, sustained or amplified biological functions.
  • Examples of enhanced T cell function include: increased secretion of y-interferon from CD8 + effector T cells, increased secretion of y-interferon from CD4 + memory and/or effector T cells, increased proliferation of CD4 + effector and/or the memory T cells, increased proliferation of CD8 + effector T cells, and increased response to antigen (e.g., clearance), when compared with the level before treatment.
  • enhancing immune response refers to stimulating, irritating, increasing, improving or enhancing the response of the immune system in mammalian.
  • the immune response can be a cellular response (i.e., cell-mediated, such as cytotoxic T lymphocyte-mediated) or a humoral response (i.e., antibody-mediated response), and can be the primary or secondary immune response.
  • cellular response i.e., cell-mediated, such as cytotoxic T lymphocyte-mediated
  • humoral response i.e., antibody-mediated response
  • Examples of enhanced immune response include: increased activity of CD4 + helper T cell and production of cytotoxic T cells.
  • the enhanced immune response can be evaluated by some in vitro or in vivo measurements known to those skilled in the art, comprising but not limited to cytotoxic T lymphocyte assay, cytokine release (such as IL-2 production), regression of tumor, and survival of tumor-carrying animals, antibody production, proliferation of immune cells, expression of cell surface marker and cytotoxicity.
  • the method enhances cellular immune response, particularly cytotoxic T cell response.
  • Tumor immunity refers to the process by which tumors escape from immune recognition and clearance. As a treatment concept, the tumor will be recognized and attacked by the immune system and consequently the patient will be treated, when tumor immunity is weakened in its ability to escape from immune recognition and clearance. Examples of tumor recognition include binding to tumor, shrinkage of tumor, and clearance of tumor.
  • Teffs T effector cells
  • T cells having cytolytic activity for example, CD4 + and CD8+T cells
  • Th cells T helper cells. Teffs secrete cytokines and activate and guide immune cells other than regulatory T cells (Treg cells).
  • the anti-OX40 antibodies described in the present disclosure can activate Teff cells, such as CD4 + and CD8 + Teff cells.
  • Treg cells mean a specialized type of CD4 + T cells that can suppress the response of other T cells.
  • Treg cells are characterized by expressing CD4, a subunit of IL-2 receptor (CD25), and the transcription factor forkhead box P3 (FOXP3) (Sakaguchi, Annu Rev Immunol 22, 531-62 (2004)), and play a vital role in the induction and maintenance of peripheral autologous tolerance. The tolerance targets against antigens expressed by tumors.
  • OX40 refers to a receptor that binds to OX40 ligand (OX40-L), and it is a member of the TNF-receptor superfamily. OX40 is also known as tumor necrosis factor receptor superfamily member 4 (TNFRSF4), ACT35, IMD16, TXGP1L and CD134.
  • TNFRSF4 tumor necrosis factor receptor superfamily member 4
  • ACT35 tumor necrosis factor receptor superfamily member 4
  • IMD16 TXGP1L
  • CD134 CD134
  • the term “OX40” includes any variant or isotype of OX40 that is naturally expressed by a cell. Therefore, the OX40 antibodies or fragment thereof described in the present disclosure can cross-react with OX40 from species other than humans (for example, cynomolgus monkey OX40). Alternatively, the OX40 antibodies or fragment thereof may be specific for human OX40, and does not exhibit cross-reactivity with OX40 of other species.
  • OX40 or variant and isoform thereof is isolated from cells or tissues in which it is naturally expressed, or recombinantly produced using techniques well known in the art and/or in the present disclosure.
  • OX40 can be natural OX40 derived from any vertebrate source, comprising mammals such as primates (e.g., humans) and rodents (e.g., mice and rats).
  • the term covers “full length”, unprocessed OX40 and any forms of OX40 due to processing occurred within a cell.
  • the term also covers naturally occurring variants of OX40, such as splicing variants or allelic variants.
  • OX40 activation refers to the activation of the OX40 receptor. Generally, OX40 activation leads to signaling.
  • an “anti-OX40 antibody” or “an antibody that binds to OX40” refers to an antibody that can bind to OX40 with sufficient affinity so that the antibody can be used as a diagnostic and/or therapeutic agent for targeting OX40.
  • reducing OX40-expressing cells means that the anti-OX40 antibody or fragment thereof kills or eliminates OX40-expressing cells. Reducing OX40-expressing cells can be achieved through various mechanisms, such as antibody-dependent cell mediated cytotoxicity (ADCC) and/or phagocytosis.
  • ADCC antibody-dependent cell mediated cytotoxicity
  • cytokine is a general term for a type of proteins that are released by a population of cells, and the cytokines act as intercellular mediators effecting on another cell.
  • cytokines are lymphokines, monocytes; interleukins (ILs), such as IL-1, IL-la, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL-12, IL-15; tumor necrosis factors, such as TNF- ⁇ or TNF- ⁇ ; and other polypeptide factors, comprising LIF and kit ligand (KL) and ⁇ -Interferon.
  • ILs interleukins
  • TNF- ⁇ or TNF- ⁇ tumor necrosis factors
  • KL kit ligand
  • cytokines involve proteins from natural sources or from recombinant cell culture, and the biologically active equivalents thereof, comprising small molecular entities produced by artificial synthesis, and pharmaceutically acceptable derivatives and salts thereof.
  • antibody is not limited by any specific method for producing antibodies. For example, it involves recombinant antibodies, monoclonal antibodies, and polyclonal antibodies.
  • the antibodies may be antibodies of different isotypes, for example, IgG (e.g., IgG1, IgG2, IgG3 or IgG4 subtype), IgA1, IgA2, IgD, IgE or IgM antibodies.
  • variable region Near the N-terminus of the antibody heavy chain and light chain, a sequence of about 110 amino acid varies largely, known as variable region (V region); the rest of the amino acid sequence near the C-terminus is relative stable, known as constant region (C region).
  • the variable region comprises three hypervariable regions (HVRs) and four framework regions (FRs) having relatively conserved sequence.
  • the three hypervariable regions determine the specificity of the antibody, also known as complementarity determining regions (CDRs).
  • Each light chain variable region (VL) and each heavy chain variable region (VH) is composed of three CDRs and four FRs, with an order from the amino terminus to the carboxyl terminus being: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • Three light chain CDRs refer to LCDR1, LCDR2, and LCDR3; three heavy chain CDRs refer to HCDR1, HCDR2 and HCDR3.
  • the number and position of the CDR amino acid residues in the LCVR region and HCVR region of the antibody or antigen-binding fragment thereof described in the present disclosure comply with the known Kabat numbering criteria (for LCDR1-3, HCDR1-3).
  • recombinant human antibody includes human antibodies prepared, expressed, created or isolated by recombinant method, and the techniques and methods involved are well known in the art, such as:
  • Such recombinant human antibodies comprise variable regions and constant regions, which involve specific human germline immunoglobulin sequences encoded by germline genes, but also involve subsequent rearrangements and mutations, such as those occured during the antibody maturation.
  • the antibodies of the present disclosure refer to murine antibodies, chimeric antibodies, humanized antibodies, and human antibodies.
  • the antibodies are humanized antibodies.
  • murine antibody herein refers to monoclonal antibodies against human OX40, which are prepared according to the knowledge and skills in the art. During the preparation, a test object is injected with OX40 antigen, and then splenocytes (B lymphocytes) expressing the antibody which possesses desired sequence or functional characteristics are separated, and then the B lymphocytes are fused with myeloma cells to obtain corresponding hybridoma cells.
  • B lymphocytes splenocytes expressing the antibody which possesses desired sequence or functional characteristics
  • the murine OX40 antibody or antigen-binding fragment thereof further comprises light chain constant region(s) of murine K, X, chain or variant(s) thereof, or further comprises heavy chain constant region(s) of murine IgG1, IgG2, IgG3 or IgG4, or variant(s) thereof.
  • human antibody includes antibodies having variable and constant region(s) from human germline immunoglobulin sequences.
  • Human antibodies of the present disclosure may include amino acid residues that are not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
  • human antibody does not refer to such an antibody in which CDR sequences derived from other mammalian species (such as mouse) germline have been grafted onto human framework sequence (i.e., “humanized antibody”).
  • humanized antibody refers to antibodies generated by grafting non-human species CDR sequences onto variable region framework of human antibody; that is, antibodies produced from different types of human germline antibody framework sequences. Humanized antibodies overcome the heterogenous response induced by chimeric antibodies which carry a large amount of heterogeneous protein components.
  • framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences.
  • the germline DNA sequences of human heavy chain and light chain variable region genes can be obtained in “VBase” human germline sequence database (www.mrccpe.com.ac.uk/vbase), and also in Kabat, E. A, et al., 1991 Sequences of Proteins of Immunological Interest, 5th edition.
  • humanized antibodies of the present disclosure also include humanized antibodies that are obtained after further affinity maturation of CDRs by phage display or yeast display.
  • the reduced affinity of the resulting OX40 antibody (or antigen-binding fragment thereof) to the antigen is due to changes occurred in the framework residues which are responsible for the contact with the antigen. Such interaction can be the result of hypermutation in somatic cells. Therefore, there is still a need to graft such donor framework amino acids onto the framework of humanized antibodies.
  • the amino acid residues from non-human OX40 antibody or antigen-binding fragment thereof involved in antigen-binding can be identified by detecting the sequence and structure of the variable region of non-human monoclonal antibody. Residues in a CDR donor framework that are different from those in the germline can be considered to be related.
  • the sequence can be aligned against the consensus sequence of a subtype or against the consensus sequence of a murine sequence with high percentage of similarity. Rare framework residues are thought to be the result of hypermutation in somatic cells and thus play an important role in binding.
  • the antibody light chain of the OX40 humanized antibody further comprises a light chain constant region of a human kappa, lambda chain or the variant thereof.
  • the antibody heavy chain of the OX40 humanized antibody further comprises a heavy chain constant region of human IgGl, IgG2, IgG3, IgG4 or the variant thereof; in particular, a human IgG1 heavy chain constant region.
  • back mutation refers to reversion of the amino acid mutation(s) in the FR region of a human antibody to the amino acid residue(s) at the corresponding position(s) of the original antibody source.
  • the variable region of the humanized antibody can be subjected to minimal back mutation(s) to maintain the activity of the antibody.
  • chimeric antibody is an antibody which is formed by fusing the variable region of a non-human antibody with the constant region of a human antibody, the chimeric antibody can alleviate the immune response induced by non-human antibody.
  • hybridoma secreting specific murine monoclonal antibody shall be established firstly, a variable region gene is then cloned from mouse hybridomas, then a constant region gene of a human antibody is cloned as desired, the mouse variable region gene is ligated to the human constant region gene to form a chimeric gene which can be inserted into a human vector, and finally the chimeric antibody molecule is expressed in an eukaryotic or prokaryotic industrial system.
  • the constant region of a human antibody is selected from heavy chain constant region(s) of human IgG1, IgG2, IgG3 or IgG4 or variant(s) thereof.
  • a chimeric antibody comprises human IgG1 heavy chain constant region(s).
  • antigen-binding fragment or “functional fragment” of an antibody refers to one or more fragments of the antibody that retain the ability to specifically bind to an antigen (e.g., OX40). It has been shown that fragments of a full-length antibody can be used to achieve function of antigen-binding. Examples of the binding fragments contained in the term “antigen-binding fragment” of an antibody involve:
  • Fab fragment a monovalent fragment consisting of VL, VH, CL and CH1 domains
  • F(ab′) 2 fragment a bivalent fragment formed by two Fab fragments connected by disulfide bridge(s) in the hinge region;
  • CDR complementarity determining region
  • VL domain and VH domain of the Fv fragment are encoded by two separate genes, however they can be linked by a synthetic linker by using recombinant methods, to generate a single protein chain in which a monovalent molecular is formed by pairing the VL with VH domain (referred to as single chain Fv (scFv); see, e.g., Bird et al. (1988): Science 242:423-426; and Huston et al (1988) Proc. Natl. Acad. Sci USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be included in the term “antigen binding fragment” of an antibody.
  • Antigen binding portions can be produced by recombinant DNA technology or by enzymatic or chemical digestion of an intact immunoglobulin.
  • Antibodies can be in the forms of different isotypes, e.g., IgG (e.g., IgG1, IgG2, IgG3 or IgG4 subtype), IgAl, IgA2, IgD, IgE or IgM antibody.
  • antigen-binding fragment refers to Fab, Fv, sFv, F(ab′) 2 , linear antibody, single-chain antibody, scFv, sdAb, sdFv, nanobody, peptibody, domain antibody and multispecific antibody (bispecific antibody, diabody, triabody and tetrabody, tandem di-scFv, tandem tri-scFv) having antigen-binding activity.
  • Fab is an antibody fragment obtained by treating an IgG antibody molecule with a papain (which cleaves the amino acid residue at position 224 in H chain).
  • the obtained fragment has a molecular weight of about 50,000 and antigen-binding activity, in which almost half of H chain to the N-terminus and the entire L chain are bound together through a disulfide bond.
  • the Fab in the present disclosure can be produced by treating the monoclonal antibody of the present disclosure (which specifically recognizes human OX40 and binds to the amino acid sequence of extracellular domain or the 3D structure thereof) with papain.
  • the Fab can be produced by inserting DNA encoding Fab of the antibody into a prokaryotic expression vector or eukaryotic expression vector and introducing the vector into a prokaryote or eukaryote to express the Fab.
  • F(ab)2 refers to an antibody fragment having a molecular weight of about 100,000 and antigen-binding activity, which is obtained by digesting the part downstream of the two disulfide bonds in the hinge region of IgG by pepsin. F(ab′) 2 contains two Fabs connected at the hinge region.
  • F(ab′) 2 of the present disclosure can be produced by treating the monoclonal antibody of the present disclosure with pepsin. Also, F(ab′) 2 can be produced by connecting the Fab′ described below via thioether bond or disulfide bond.
  • Fab′ is an antibody fragment having a molecular weight of about 50,000 and having antigen-binding activity, which is obtained by cleaving the disulfide bond at the hinge region of the above-mentioned F(ab′) 2 .
  • the Fab′ of the present disclosure can be produced by treating F(ab′) 2 of the present disclosure with a reducing agent such as dithiothreitol.
  • the Fab′ can be produced by inserting DNA encoding Fab′ of the antibody into a prokaryotic expression vector or eukaryotic expression vector and introducing the vector into a prokaryote or eukaryote to express the Fab′.
  • single chain antibody refers to a molecule comprising antibody heavy chain variable domain (or region; VH) connected to antibody light chain variable domain (or region; VL) by a linker.
  • Such scFv molecules have general structure of NH 2 -VL-linker-VH—COOH or NH 2 -VH-linker-VL-COOH.
  • a suitable linker in the prior art consists of repeated GGGGS amino acid sequence or variant thereof, for example, variant with 1-4 repeats (Holliger et al. (1993), Proc. Natl. Acad. Sci. USA 90:6444-6448).
  • linkers that can be used in the present disclosure are described by Alfthan et al. (1995), Protein Eng. 8:725-731, Choi et al. (2001), Eur. J. Immunol. 31:94-106, Hu et al. (1996), Cancer Res. 56:3055-3061, Kipriyanov et al. (1999), J. Mol. Biol. 293:41-56 and Roovers et al. (2001), Cancer Immunol.
  • the scFv of the present disclosure can be produced by the following steps: obtaining cDNAs encoding the VH and VL of the monoclonal antibody of the present disclosure, constructing a DNA encoding the scFv, inserting the DNA into a prokaryotic or eukaryotic expression vector, and then introducing the expression vector into a prokaryote or eukaryote to express the scFv.
  • Diabody is an antibody fragment in which the scFv is dimerized, and it is an antibody fragment having divalent antigen-binding activity. In the divalent antigen-binding activity, the two antigens may be the same or different.
  • the diabody of the present disclosure can be produced by the following steps: obtaining cDNAs encoding VH and VL of the monoclonal antibody of the present disclosure, constructing a DNA encoding scFv to make the length of a linker peptide being of 8 or less amino acid residues, inserting the DNA into a prokaryotic or eukaryotic expression vector, and then introducing the expression vector into a prokaryote or eukaryote to express the di ab ody.
  • “dsFv” is obtained by replacing one amino acid residue in each of VH and VL with a cysteine residue, and then connecting the substituted polypeptides via a disulfide bond between the two cysteine residues.
  • the amino acid residues to be replaced with a cysteine residue can be selected based on three-dimensional structure prediction of the antibody in accordance with known methods (Protein Engineering, 7, 697 (1994)).
  • the dsFv of the present disclosure can be produced by the following steps: obtaining cDNAs encoding the VH and VL of the monoclonal antibody of the present disclosure, constructing a DNA encoding the dsFv, inserting the DNA into a prokaryotic or eukaryotic expression vector, and then introducing the expression vector into a prokaryote or eukaryote to express the dsFv.
  • CDR-containing peptide is constituted by incorporating one or more regions in the CDRs of VH or VL. Peptides containing multiple CDRs can be linked directly or via a suitable peptide linker.
  • the CDR-containing peptide of the present disclosure can be produced by the following steps: constructing DNA encoding the CDRs of VH and VL of the monoclonal antibody in the present disclosure, inserting the DNA into a prokaryotic expression vector or a eukaryotic expression vector, the expression vector is then introduced into prokaryotes or eukaryotes to express the peptide.
  • the CDR-containing peptide can also be produced by a chemical synthesis method, such as Fmoc method or tBoc method.
  • FR framework
  • VL variable domain
  • VH variable domain
  • CDRs antigen binding loops
  • amino acid difference refers to the difference(s) between a polypeptide and the variant(s) thereof at specific amino acid position(s) on the polypeptide fragment, wherein the variant(s) can be obtained by substituting, inserting or deleting amino acid(s) at specific position(s) on the polypeptide.
  • epitope refers to the sites on an antigen that specifically bind to an immunoglobulin or antibody.
  • the epitope can be formed by adjacent amino acids, or by non-adjacent amino acids which have been brought to be closer due to tertiary folding of a protein.
  • the epitope formed by adjacent amino acids is typically retained upon exposure to denaturing solvent, whereas the epitope formed by tertiary folding is typically absent after treatment with denaturing solvent.
  • Epitopes typically include at least 3-15 amino acids in a unique spatial conformation.
  • Methods for determining epitope are well known in the art, comprising immune-blotting and immune-precipitation assay, and the like. Methods for determining the spatial conformation of an epitope include techniques in the art and techniques described herein, such as X-ray crystallography and two-dimensional nuclear magnetic resonance, and the like.
  • the term “specifically binds to” used in present invention refers to the binding of an antibody to an epitope on a predetermined antigen.
  • the antibody binds to a predetermined antigen with an equilibrium dissociation constant (KD) of approximately less than 10′ M or even less, and the affinity of the antibody for binding to the predetermined antigen is at least two times higher than that for binding to non-specific antigens (such as BSA) other than the predetermined antigen or closely-related antigens, as measured in an instrument via surface plasmon resonance (SPR) technique, wherein the recombinant human OX40 is used as an analyte while the antibody is used as a ligand.
  • SPR surface plasmon resonance
  • an antibody recognizing an antigen can be used interchangeably herein with the term “an antibody specifically binding to.”
  • KD refers to the dissociation equilibrium constant for particular antibody-antigen interaction.
  • the antibody of the present disclosure binds to human OX40 with a dissociation equilibrium constant (K D ) of less than about 10′M, for example, less than about 10 ⁇ 8 M, 10 ⁇ 9 M or 10 ⁇ 10 M or even less; for example, as determined by Surface Plasma Resonance (SPR) technology in Biacore instrument.
  • SPR Surface Plasma Resonance
  • competition occurs between the antigen-binding proteins, which is determined by an assay in which: an antigen binding protein to be tested (e.g., an antibody or an antigen binding fragment thereof) prevents or inhibits (e.g., reduces) the specific binding of a reference antigen binding protein (e.g., a ligand or reference antibody) to a common antigen (e.g., a OX40 antigen or fragment thereof).
  • an antigen binding protein to be tested e.g., an antibody or an antigen binding fragment thereof
  • a reference antigen binding protein e.g., a ligand or reference antibody
  • a common antigen e.g., a OX40 antigen or fragment thereof.
  • assays are, for example, solid phase direct or indirect radioimmunoassay (MA), solid phase direct or indirect enzyme immunoassay (EIA), Sandwich competition assay (see, e.g., Stahli et al, 1983, Methods in Enzymology 9: 242-253); solid phase direct biotin-avidin EIA (see, e.g., Kirkland et al, 1986, J. Immunol.
  • MA solid phase direct or indirect radioimmunoassay
  • EIA enzyme immunoassay
  • Sandwich competition assay see, e.g., Stahli et al, 1983, Methods in Enzymology 9: 242-253
  • solid phase direct biotin-avidin EIA see, e.g., Kirkland et al, 1986, J. Immunol.
  • solid phase direct labeling assay solid phase direct labeling sandwich assay (see, e.g., Harlow and Lane, 1988, Antibodies, A Laboratory Manual, Cold Spring Harbor Press); solid phase direct labeling RIA with I-125 label (see, e.g., Morel et al, 1988, Molec. Immunol. 25: 7-15); and direct labeling RIA (Moldenhauer et al, 1990, Scand. J. Immunol. 32: 77-82).
  • the assay involves the use of a purified antigen capable of binding to both an unlabeled test antigen binding protein and a labeled reference antigen binding protein (the antigen is loaded on a solid surface or cell surface).
  • Antigen-binding proteins identified by competitive assay includes: antigen-binding proteins that bind to the same epitope as the reference antigen-binding protein; and antigen-binding proteins that bind to an epitope that is sufficiently close to the epitope to which the reference antigen-binding protein binds, where the two epitopes spatially interfere with each other to hinder the binding. Additional details regarding methods for determining competitive binding are provided in the examples herein.
  • a competitive antigen-binding protein when present in excess, it will inhibit (e.g., reduce) at least 40-45%, 45-50%, 50-55%, 55-60%, 60-65%, 65-70%, 70-75% or even more of the specific binding of the reference antigen-binding protein to the common antigen. In certain cases, the binding is inhibited by at least 80-85%, 85-90%, 90-95%, 95-97% or even more.
  • cross-reaction refers to the ability of the antibody of the present invention to bind to OX40 from different species.
  • an antibody of the present invention that binds to human OX40 can also bind to OX40 from another species.
  • Cross-reactivity is measured by detecting the specific reactivity with purified antigen in binding assays (e.g., SPR and ELISA), or by detecting the binding or functional interaction with cells physiologically expressing OX40.
  • binding assays e.g., SPR and ELISA
  • Methods for determining cross-reactivity include standard binding assays as described herein, such as surface plasmon resonance analysis (SPR), or flow cytometry.
  • inhibitortion or “blocking” are used interchangeably, and covers both partial and complete inhibition/blocking.
  • inhibition of growth e.g., when applied to cells
  • inhibition of growth is intended to refer to any measurable decrease in cell growth.
  • inducing an immune response and “enhancing an immune response” are used interchangeably, and refer to an immune response against the stimulation of a specific antigen (i.e, passive or adaptive).
  • induction in the context of inducing CDC or ADCC refers to the stimulation of a specific direct cell killing mechanism.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • FcR Fc receptors
  • cytotoxic cells such as NK cells, neutrophils, and macrophages
  • NK cells The main cells that mediate ADCC express FcyRIII alone, while monocytes express FcyRI, FcyRII, and FcyRIII.
  • FcR expression on hematopoietic cells is summarized in Table 3 on page 464 in Ravetch and Kinet, Annu. Rev. Immunol.
  • an in vitro ADCC assay can be performed, such as that described in U.S. Pat. No. 5,500,362 or U.S. Pat. No. 5,821,337 or U.S. Pat. No. 6,737,056 (Presta).
  • the effector cells useful for such assays include PBMC and NK cells.
  • the ADCC activity of the molecule of interest can be assessed in vivo, for example in an animal model such as that disclosed in Clynes et al. PNAS (USA) 95:652-656 (1998).
  • the Fc segment of IgG can be modified to reduce or eliminate the ADCC effect of antibodies.
  • the modification refers to mutations in the heavy chain constant region of the antibody, such as mutations selected from the group consisting of N297A, L234A, L235A of IgG1; IgG2/4 chimera, F235E of IgG4, and L234A/E235A mutation.
  • complement dependent cytotoxicity refers to the lysis of target cells in the presence of complement.
  • the classical pathway of complement activation is initiated by the binding of the first component of the complement system (C1q) to an antibody (of the appropriate subclass), which has bound to its cognate antigen.
  • C1q first component of the complement system
  • an antibody of the appropriate subclass
  • a CDC assay can be performed, for example as described in Gazzano-Santoro et al., J. Immunol. Methods 202:163 (1996).
  • Polypeptide variants having modified Fc region amino acid sequence(s) (polypeptides with variant Fc region(s)) as well as having increased or decreased C1q-binding ability are described in, for example, U.S. Pat. No. 6,194,551B1 and WO 1999/51642. See also, for example, Idusogie et al., J. Immunol. 164: 4178-4184 (2000).
  • nucleic acid molecule refers to DNA molecule and RNA molecule.
  • a nucleic acid molecule may be a single-stranded or double-stranded, but in particular double-stranded DNA.
  • a nucleic acid is considered to be “operably linked”, when it is placed into a functional relationship with another nucleic acid sequence. For instance, a promoter or enhancer is operably linked to an encoding sequence, when it has effect on the transcription of the sequence.
  • vector refers to a nucleic acid molecule capable of delivering another nucleic acid to which it has been linked.
  • the vector is a “plasmid,” which refers to a cyclic double-stranded DNA loop into which additional DNA segment(s) may be ligated.
  • the vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • the vectors disclosed herein are capable of self-replicating in the host cell into which they are introduced (e.g., bacterial vectors having a bacterial replication origin and episomal mammalian vectors), or may be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the genome of the host (e.g., non-episomal mammalian vectors).
  • mice can be immunized with human OX40 or fragments thereof, and the resulting antibodies can then be renatured, purified, and sequenced for amino acid sequences by using conventional methods well known in the art.
  • Antigen-binding fragments can also be prepared by conventional methods.
  • the antibodies or antigen binding fragments thereof of the present disclosure are engineered to insert one or more human FR regions onto non-human CDR regions.
  • Human FR germline sequences can be obtained from ImMunoGeneTics (IMGT) website http://imgt.cines.fr, or from The Immunoglobulin Facts Book, 2001 ISBN012441351, by aligning against the IMGT human antibody variable germline gene database using MOE software.
  • IMGT ImMunoGeneTics
  • the antibodies or the antigen-binding fragments in the present disclosure may be prepared and purified using conventional methods.
  • cDNA sequences encoding a heavy chain and a light chain may be cloned and recombined into GS expression vector.
  • the recombinant immunoglobulin expression vector then may be stably transfected into CHO cells.
  • mammalian expression system may result in glycosylation of antibodies, typically at the highly conserved N-terminus in the Fc region.
  • Stable clones may be obtained through expression of an antibody specifically binding to human antigen. Positive clones may be expanded in a serum-free culture medium for antibody production in bioreactors.
  • Culture medium into which an antibody has been secreted, may be purified and collected by conventional techniques.
  • the antibody may be subjected to filtration and concentration using common techniques. Soluble mixtures and multimers may be effectively removed by common techniques, such as molecular sieve or ion exchange.
  • the obtained product shall be immediately frozen, for example at ⁇ 70° C., or may be lyophilized.
  • the monoclonal antibody refers to an antibody obtained from a single clone of cell strain which is but not limited to eukaryotic, prokaryotic, or phage clone of cell strain.
  • Monoclonal antibodies and antigen-binding fragment thereof can be obtained, for example, by hybridoma technologies, recombinant technologies, phage display technologies, synthetic technologies (e.g., CDR-grafting), or other technologies known in the art.
  • host cell refers to a cell into which an expression vector has been introduced.
  • Host cells may include microorganisms (such as bacteria), plants or animal cells.
  • Bacteria susceptible to be transformed include members of the Enterobacteriaceae, such as Escherichia coli or Salmonella; Bacillaceae, such as Bacillus subtilis; Pneumococcus; Streptococcus and Haemophilus influenzae.
  • Suitable microorganisms include Saccharomyces cerevisiae and Pichia pastoris.
  • Suitable animal host cell lines include CHO (Chinese Hamster Ovary Cell Line), NSO and 293 cells.
  • Constant modification or “conservative replacement or substitution” means the substitution of other amino acids showing similar characteristics (such as charge, side chain size, hydrophobicity/hydrophilicity, main chain conformation and rigidity, etc) for the amino acids in a protein, such that the modification can be frequently performed without changing the biological activity of the protein.
  • Those skilled in the art know that, generally, single amino acid substitution in a non-essential region of a polypeptide does not substantially change the biological activity (see for example, Watson et al. (1987) Molecular Biology of the Gene, The Benjamin/Cummings Pub. Co., Page 224 (4th edition)).
  • the substitution of amino acids with similar structure or function is unlikely to result in the loss of biological activity.
  • Effective amount involves an amount sufficiently to ameliorate or prevent a symptom or sign of a medical symptom or condition. Effective amount also means an amount sufficiently to allow or facilitate diagnosis. An effective amount for a particular patient or veterinary subject may vary depending on factors such as the condition being treated, the general health of the patient, the route and dosage of administration and the severity of side effects. An effective amount can be the maximal dosage or dosing protocol that avoids significant side effects or toxic effects.
  • Exogenous refers to substances that are produced outside an organism, cell, or human body, depending on the context.
  • Endogenous refers to substances that are produced within an organism, cell, or human body, depending on the context.
  • the “mutated sequence” mentioned in the present disclosure refers to a nucleotide sequence and amino acid sequence having various percentage sequence identity to those of the present disclosure, after modifying the nucleotide sequence and amino acid sequence of the present disclosure by appropriate substitution, insertion or deletion.
  • the sequence identity may be at least 85%, 90% or 95%, non-limiting examples include 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and 100%. Sequence comparison between two sequences and determination of identity percent can be performed using the BLASTN/BLASTP algorithm available on the National Center For Biotechnology Institute website, with default settings.
  • homology refers to a sequence similarity between two polynucleotide sequences or between two polypeptide sequences.
  • a position in both of the two sequences to be compared is occupied by the same base or amino acid monomer subunit, e.g., when a position in each of two DNA molecules is occupied by adenine, then the molecules are homologous at that position.
  • the percent of homology between two sequences is a function of the number of matching or homologous positions shared by the two sequences divided by the number of all positions to be compared and then multiplied by 100.
  • the expressions “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations involve progeny thereof.
  • “transformant” and “transformed cell” include the primary subject cells and culture derived therefrom, regardless of the number of passages. It should be also understood that all progeny may not be precisely identical in the aspect of DNA content, due to intended or unintended mutations. Mutant progeny exhibiting the same function or biological activity as screened for the originally transformed cells are included. Where distinct designations are mentioned, it will be clearly understood according to the context.
  • PCR polymerase chain reaction
  • sequence information at the terminus of or beyond the region of interest needs to be available, such that oligonucleotide primers can be designed; these primers will be identical or similar in sequence to the complementary strand of the template to be amplified.
  • the 5′ terminus nucleotides of the two primers can be identical to the ends of the material to be amplified.
  • PCR can be used to amplify specific RNA sequences, specific DNA sequences from total genome and cDNA transcribed from total cellular RNA, bacteriophage or plasmid sequences, etc. See generally Mullis et al. (1987) Cold Spring Harbor Symp. Ouant. Biol. 51:263; Erlich editor, (1989) PCR TECHNOLOGY (Stockton Press, NY).
  • the PCR used in the present disclosure is considered to be one example (but not the only one) of polymerase reaction methods for amplifying a nucleic acid sample to be testes.
  • the method comprises the use of nucleic acid sequences known as primers in combination with nucleic acid polymerase to amplify or generate a specific portion of nucleic acid.
  • “Optional” or “optionally” means that the event or circumstance that follows may (but does not necessarily) occur, and the description includes the instances where the event or circumstance does or does not occur.
  • “optionally contains 1-3 antibody heavy chain variable region(s)” means the antibody heavy chain variable region(s) with specific sequence can be, but need not be, present.
  • “Pharmaceutical composition” refers to a mixture containing one or more antibodies or the antigen binding fragments thereof according to the present disclosure, and other chemical components, such as physiologically/pharmaceutically acceptable carriers or excipients.
  • the pharmaceutical composition aims at promoting the administration to an organism, facilitating the absorption of the active component and thereby exhibiting a biological effect.
  • cancer refers to or describes a physiological condition in mammals, characterized by unregulated cell growth.
  • examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • cancers include, but are not limited to, squamous cell carcinoma (e.g., epithelial squamous cell carcinoma), lung cancer (including small cell lung cancer, non-small cell lung cancer, adenocarcinoma, and squamous cell carcinoma of the lung), peritoneal cancer, hepatocellular carcinoma, gastric carcinoma (including gastrointestinal cancer and gastrointestinal stromal cancer), pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, urethral cancer, liver tumor, breast cancer, colon cancer, rectum cancer, colorectal cancer, endometrial cancer or uterine cancer, salivary gland cancer, kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, anal cancer, penile cancer, melanoma, superficial spreading melanoma, malignant freckle mole melanoma, acral melanoma, nodular melanoma, multiple myeloma and B-cell lympho
  • cancers suitable to be treated by the OX40 antibody or the fragment thereof in the present disclosure include breast cancer, colorectal cancer, rectal cancer, non-small cell lung cancer, glioblastoma, non-Hodgkin's lymph Tumor (NHL), renal cell carcinoma, prostate cancer, liver cancer, pancreatic cancer, soft tissue sarcoma, Kaposi's sarcoma, carcinoid carcinoma, head and neck cancer, ovarian cancer, mesothelioma, and multiple Myeloma.
  • the cancer is selected from the group consisting of: non-small cell lung cancer, glioblastoma, neuroblastoma, melanoma, breast cancer (e.g.
  • cancer is selected from the group consisting of: non-small cell lung cancer, colorectal cancer, glioblastoma, and breast cancer (e.g. triple negative breast cancer), including metastases of those cancers.
  • cell proliferative disorder and “proliferative disorder” refer to disorders associated with a certain degree of abnormal cell proliferation. In one embodiment, the cell proliferative disorder refers to cancer.
  • tumor refers to all neoplastic cell growth and proliferation, regardless of malignant or benign, and all pre-cancerous and cancerous cells and tissues.
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • cancer cancer
  • administering when applied to an animal, human, subject, cell, tissue, organ or biological fluid, refers to contacting an exogenous pharmaceutical, therapeutic, diagnostic agent, or composition with the animal, human, subject, cell, tissue, organ or biological fluid.
  • administering can refer to such as therapeutic, pharmacokinetic, diagnostic, research and experimental methods. Treatment of a cell involves contacting a reagent with a cell, as well as contacting a reagent with a fluid wherein said fluid is in contact with the cell.
  • administering also mean in vitro and ex vivo treatment of e.g. a cell, by using a reagent, diagnostic, binding composition, or by using another cell.
  • Treatment when applied to a human, veterinary or a research subject, refers to therapeutic treatment, prophylactic or preventative measures, research as well as diagnostic applications.
  • “To treat” means internal or external administration of a therapeutic agent (such as a composition comprising any of the antibodies or antigen-binding fragment thereof of the present disclosure; or a nucleic acid molecule encoding the antibody or antigen-binding fragment thereof) to a patient having one or more disease or symptom for which the therapeutic agent was known to show therapeutic activity.
  • a therapeutic agent such as a composition comprising any of the antibodies or antigen-binding fragment thereof of the present disclosure; or a nucleic acid molecule encoding the antibody or antigen-binding fragment thereof
  • the therapeutic agent is administered in an amount effective to alleviate one or more disease or symptom in the treated patient or population, either by inducing the regression of such symptom(s), or by inhibiting the progression of such symptom(s) to any clinically measurable degree.
  • the amount of a therapeutic agent that is effective to alleviate any particular disease or symptom may vary according to factors such as the disease state, age and weight of the patient, and the ability of the medicament to elicit a desired effect in the patient. Whether a disease symptom has been alleviated can be assessed by any clinical measurement typically used by physicians or other skilled healthcare providers to assess the severity or progression status of that symptom.
  • the embodiment of the present disclosure may not be effective in alleviating each disease symptom of interest, it should alleviate the target disease symptom(s) of interest in a statistically significant number of subjects as determined by any statistical test known in the art such as Student's t-test, chi-square test, U-test according to Mann and Whitney, Kruskal-Wallis test (H-test), Jonckheere-Terpstra-test and Wilcoxon-test.
  • any statistical test known in the art such as Student's t-test, chi-square test, U-test according to Mann and Whitney, Kruskal-Wallis test (H-test), Jonckheere-Terpstra-test and Wilcoxon-test.
  • prevention of cancer refers to delaying, inhibiting or preventing the onset of cancer in mammals; the initiation of the development of cancer or tumor has not been confirmed in the mammals, but the mammals have been identified to be susceptible to the cancer by for example genetic screening or other methods.
  • the term also includes the treatment of a mammal suffering from precancerous condition in order to prevent the precancerous condition from progressing towards malignant tumor or even to achieve regression.
  • the library of anti-human OX40 monoclonal antibodies was generated by immunizing mice.
  • Experimental mice of the BalB/C and A1J strains Center for Comparative Medicine, Yangzhou University, Animal Production License Number; SOCK(Jiangsu)2017-007), female, 10-week old.
  • Immune antigen was human OX40 recombinant protein with Fc tag (OX40-Fc: OX40 Leu29-Ala216 (Accession# NP 003318), fused with Fc), and was purchased from Acro Biosystems under catalog number # OX40-H5255, expressed in HEK293, and then purified according to conventional method.
  • OX40-Fc was emulsified with Freund's adjuvant: Freund's complete adjuvant (sigma-aldrich, F5881-10ML) was used for the first immunization, and Freund's incomplete adjuvant (sigma-aldrich, F5506-10ML) was used for the reset of booster immunizations.
  • the ratio of antigen to adjuvant was 1:1, and 25 ⁇ g protein/200 ⁇ l/mouse was injected for each immunization.
  • the antibody titer in mouse serum and the neutralizing activity of blocking the binding of OX40/0X4OL were determined with mouse serum by ELISA method as described in Example 2.
  • the mice with strong serum titer, affinity and ability for blocking ligand binding were selected for a final immunization and then were sacrificed.
  • the spleen cells were fused with SP2/0 myeloma cells (ATCC® CRL-1581TM) and innoculated onto a plate to obtain hybridomas.
  • Target hybridomas were selected by indirect ELISA, capture ELISA and cell-based functional screening as shown in Example 2, and monoclonal antibody strains were established by limited dilution method.
  • the established 19 strains of OX40 mouse monoclonal antibodies were produced by serum-free expression, and purified mouse monoclonal antibodies were obtained by protein A affinity chromatography.
  • Hybridoma cells secreting activated anti-OX40 antibody were selected by indirect ELISA, capture ELISA and cell functional activity screening.
  • the brief procedure of functional screening was as follows: GS-H2/OX40 stable cell line was cultivated (purchased from Genscript, cat# M00608). Diluted antibody to be tested and OX4OL (Sino Biological, 13127-H04H) solution were prepared, and added into GS-H2/OX40 cells at logarithmic growth phase.
  • RNAs were extracted from cells by conventional RNA extraction technology, and then the PCR products of the variable regions of the monoclonal antibodies were obtained by reverse transcription polymerase chain reaction (RT-PCR). The PCR products were resolved and recovered by agarose gel, then cloned into a gene vector which was then transformed into E.coli. Several transformed colonies were randomly selected, and the variable regions of monoclonal antibodies were amplified by PCR for gene sequencing. The corresponding sequences of the obtained exemplary murine monoclonal antibodies are shown below.
  • the heavy and light variable region sequences of the murine monoclonal antibody m4B5 are as follows:
  • m4B5 heavy chain variable region SEQ ID NO: 1 QVQLKQSGPGLVQPSQSLSITCTVSGFSLTSYGLHWFRQSPGKGLEWLGV IWSGGSTDYNAAFISRLSISKDDSKSQVFFKMNSLQADDTAIYYCAREEY DVWGTGTTVTVSS; m4B5 light chain variable region: SEQ ID NO: 2 DIQMTQTASSLSASLGDRVTISCRASQDISNYLNWYQQKPDGTVKLLIYY TSRLHSGVPSRFSGSGSGTDYSLTISNLEQEDVATYFCQQGNTLPWTFGG GTKLEIK.
  • the heavy and light chain variable region sequences of the murine monoclonal antibody m2G3 are as follows:
  • m2G3 heavy chain variable region SEQ ID NO: 9 QVQLKESGPGLVASSQSLSITCTVSGFSLSRYSVHWVRQPPGKGLEWLGM IWDGGNTDYNSALKSRLSISKDNSKSQVFLKMNSLQTDDTAMYYCARNPL YFSYAMDYWGQGTSVTVSS; m2G3 light chain variable region: SEQ ID NO: 10 DIQMTQSTSSLSASLGDRVTISCRASQDISNYLNWYQQKPDGTVKLLIIY TSRLQSGVPSRFSGSGSGTDYSLTISNLEQEDIATYFCQQVNTFPFTFGS GTKLEIK.
  • 20xcoating buffer was diluted to lx by using deionized water.
  • 100 ⁇ L of human OX40-His antigen (Acro biosytems, OXL-H52Q8) prepared by using 1 xcoating buffer (carbonate buffer) to a final concentration of 2 ⁇ g/mL was added to each well, and incubated at 4° C. overnight or 37° C. for 2 h.
  • the plate was washed once with PBST, 200 ⁇ L of blocking solution (PBST containing 5% skimmed milk) was added to each well, incubated at 37° C. for 2 h, and the plate was washed for 4 times with PB ST.
  • PBST blocking solution
  • the antibody to be tested was 5-fold diluted from the concentration of 10000 ng/ml, resulting in seven gradients, namely 10000 ng/ml, 2000 ng/ml, 400 ng/ml, 80 ng/ml, 16 ng/ml, 3.2 ng/ml, and 0.64 ng/ml, blank wells merely contained dilution solution, i.e., 2.5% skimmed milk in PBST), and incubated at 37° C. for 40 minutes.
  • dilution solution i.e., 2.5% skimmed milk in PBST
  • the plate was washed for 4 times with PBST, and the enzyme-labeled secondary antibody (HRP-labeled goat anti-mouse IgG, purchased from Jackson Immunoresearch, Cat# 115036071 or HRP-labeled goat anti-human IgG, purchased from Jackson Immunoresearch, Cat# 109036098) was diluted with PBST buffer; 100 ⁇ L was added to each well and incubated at 37° C. for 40min. The plate was washed for 4 times with PBST, and 100 ⁇ L TMB developing solution was added to each well, incubated for 3-15 minutes in dark at room temperature, and 50p1 stop solution (1M sulfuric acid) was added to each well. The parameters were set for the microplate reader, the OD value was read at 450-630nm, and the test data were saved.
  • HRP-labeled goat anti-mouse IgG purchased from Jackson Immunoresearch, Cat# 115036071 or HRP-labeled goat anti-human IgG, purchased from
  • 20xPBS buffer was diluted to 1 xwith deionized water.
  • 100 ⁇ L of GAM secondary antibody (Jackson Immunoresearch, 115-006-071) prepared with 1 xPBS to a final concentration of 2 ⁇ g/mL was added to each well, and incubated at 4° C. overnight or 37° C. for 2 h; the plate was washed once with PBST, 200 ⁇ L blocking solution (PBST containing 5% skimmed milk) was added to each well, incubated at 37° C. for 2 h, and the plate was washed with PBST for 4 times.
  • GAM secondary antibody Jackson Immunoresearch, 115-006-071
  • the antibody to be tested was 5-fold diluted from the concentration of 10000 ng/ml, resulting in seven gradients, namely 10000 ng/ml, 2000 ng/ml, 400 ng/ml, 80 ng/ml, 16 ng/ml, 3.2 ng/ml, and 0.64 ng/ml, blank wells merely contained dilution solution, i.e., 2.5% skimmed milk in PBST), and incubated at 37° C. for 40 minutes.
  • dilution solution i.e., 2.5% skimmed milk in PBST
  • the plate was washed for 4 times with PBST, human OX40-FC-biotin (Acro biosystem, OXO-H5255, labeled biotin) was diluted with 2.5% skimmed milk in PBST, 100 ⁇ l was added to each well, incubated at 37° C. for 40 min, and the plate was washed for 4 times with PB ST. 100 ⁇ L TMB developing solution was added to each well, incubated for 3-15 minutes in dark at room temperature, 50 ⁇ l stop solution (1M sulfuric acid) was added to each well. The parameters were set for the microplate reader, the OD value was read at 450-630nm, and the test data were saved.
  • human OX40-FC-biotin Acro biosystem, OXO-H5255, labeled biotin
  • Blocking ELISA of ligand binding 20 ⁇ coating buffer was diluted to lx by using deionized water, 100 ⁇ L of OX40L-His antigen (Acro biosytems, OXL-H52Q8) prepared by using 1 ⁇ coating buffer (carbonate buffer) to a final concentration of 2 ⁇ g/mL was added to each well, and incubated at 4° C. overnight or 37° C. for 2 h. The plate was washed once with PBST, 200 ⁇ L of blocking solution (PBST containing 5% skimmed milk) was added to each well, incubated at 37° C.
  • OX40L-His antigen Acro biosytems, OXL-H52Q8
  • mice serum/antibody was gradient-diluted with pre-prepared 200 ng/ml human OX40-Fc solution (prepared in 2.5% skimmed milk), and then pre-incubated at room temperature for 40 minutes, then added to the blocked OX4OL plate at 100 ⁇ L/well, and incubated for 40min; the plate was washed for 4 times with PBST; HRP-labeled goat anti-human secondary antibody (GAH-HRP, Jackson Immunoresearch, 109-035-006) was diluted with PBST buffer; 100 ⁇ L was added to each well and incubated at 37° C. for 40min.
  • GSH-HRP Jackson Immunoresearch, 109-035-006
  • the plate was washed for 4 times with PBST, and 100 ⁇ L TMB developing solution was added to each well, incubated for 3-15 minutes in dark at room temperature, and 50 ⁇ l stop solution (1M sulfuric acid) was added to each well.
  • the parameters were set for the microplate reader, the OD value was read at 450-630nm, and the test data were saved.
  • the murine antibody m2G3 and m4B5 heavy chain variable region (VH) in combination with human immunoglobulin heavy chain constant region, and light chain variable region (VL) in combination with human immunoglobulin Kappa light chain constant region were separately cloned into eukaryotic expression vector, which was then transfected into cells to produce mouse-human chimeric antibodies.
  • the heavy chain vector was designed as follows: signal peptide+heavy chain variable region sequence+human IgG1 constant region sequence.
  • the light chain vector was designed as follows: signal peptide +light chain variable region sequence +human Kappa constant region sequence.
  • the above sequences were inserted into pCEP4 vector (Thermofisher, V04450). After obtaining the vector plasmid, the plasmid was extracted and was delivered to sequencing for verification. The qualified plasmid was transfected into human 293F cells with PEI and cultured continuously, and the 293F cells were cultivated in serum-free medium (Shanghai OPM biosciences, OPM-293 CD03) to reach logarithmic growth phase for cell transfection.
  • chimeric antibody light chain plasmid and 23.6 ⁇ g of chimeric antibody heavy chain plasmid were dissolved in 10 ml Opti-MEM(IDI Reduced Serum Medium (GIBCO, 31985-070) and mixed well, then 200m PEI was added, mixed well, and incubated at room temperature for 15min, and added into 50mL cells.
  • Cell culture conditions 5% CO2, 37° C., 125 rpm/min. During the culture period, medium supplementary was added on day 1 and day 3 until the cell viability was less than 70%, and the cell supernatant was collected and centrifuged.
  • the cell culture solution was loaded onto affinity column for antibody purification, the column was washed with phosphate buffer, eluted with glycine-hydrochloric acid buffer (pH 2.7, 0.1M Gly-HCl), neutralized with 1M Tris hydrochloric acid pH 9.0, and dialyzed against phosphate buffer, and purified chimeric antibodies Ch2G3 and Ch4B5 were finally obtained.
  • m2G3-NC (-NC represents negative control), Ch2G3-NC, m4B5-NC and Ch4B5-NC were negative controls, which share the same constant regions as that used in m2G3, Ch2G3, m4B5, and Ch4B5, respectively, however, the variable regions thereof do not recognize OX40.
  • the well-plate was coated with anti-CD3 antibody (Chempartner, A05-001), placed at 4° C. overnight and washed for 3 times with PBS; Jurkat-NF-Kb luc-hOX40 cells (ATCC, TIB-152 stable cell line constructed by Shanghai ChemPartner) and Raji cells (ATCC, CCL-86) were harvested, resuspended and mixed. 50 ⁇ l/well of the diluted antibody to be tested and 50 ⁇ l/well of the mixed two types of cells were added into the plate, and the plate was incubated at 37° C. in 5% CO 2 incubator for 5 hours.
  • VH heavy chain variable region
  • VL light chain variable region
  • the heavy chain vector was designed as follows: signal peptide+mutated heavy chain variable region sequence+human IgG1 constant region sequence.
  • the light chain vector was designed as follows: signal peptide +mutated light chain variable region sequence +human Kappa constant region sequence.
  • the above sequences were separately inserted into pCEP4 vector (Thermofisher, V04450).
  • the expression vectors were synthesized by a third-party gene synthesis company according to the above design. After obtaining the vector plasmid, the plasmid was extracted and sent to sequencing for verification. The qualified plasmid was transfected into human 293F cells with PEI and cultured continuously, and the 293F cells were cultivated in serum-free medium (Shanghai OPM biosciences, OPM-293 CD03) to reach logarithmic growth phase for cell transfection.
  • Cell culture conditions 5% CO 2 , 37° C., 125 rpm/min. During the culture period, medium supplementary was added on day 1 and day 3 until the cell viability was less than 70%, and the cell supernatant was collected and centrifuged. After centrifugation and filtration, the cell culture solution was loaded onto affinity column for antibody purification, the column was washed with phosphate buffer, eluted with glycine-hydrochloric acid buffer (pH 2.7, 0.1M Gly-HCl), neutralized with 1M Tris hydrochloric acid pH 9.0, and dialyzed against phosphate buffer, and purified humanized antibodies were finally obtained.
  • sequences of the humanized variable regions are as follows:
  • sequences of the humanized variable regions of m4B5 are as follows:
  • hu4B5 VL0 SEQ ID NO: 21 DIQMTQSPSSLSASVGDRVTITC RASQDISNYLN WYQQKPGKVPKLLIY Y TSRLHS GVPSRFSGSGSGTDFTLTISSLQPEDVATYYC QQGNTLPWT FGG GTKVEIK ;
  • hu4B5 VL1 SEQ ID NO: 22 DIQMTQSPSSLSASVGDRVTITC RASQDISNYLN WYQQKPGKVPKLLIY Y TSRLHS GVPSRFSGSGSGTDYTLTISSLQPEDVATYYC QQGNTLPWT FGG GTKVEIK ;
  • hu4B5 VL2 SEQ ID NO: 23 DIQMTQSPSSLSASVGDRVTITC RASQDISNYLN WYQQKPGGVPKLLIY Y TSRLHS GVPSRFSGSGSGTDYTLTISSLQPEDVATYYC QQG
  • hu2G3 VH1.1 SEQ ID NO: 31 QVQLQESGPGLVKPSETLSLTCTVSGYSIS RYSVH WVRQPPGKGLEWIG M IWDGGNTDYNAALKS RFTISRDNAKNTLYLQMNSLRAEDTAVYYCAR NPL YFSYAMDY WGQGTLVTVSS; >hu2G3 VH1.2 SEQ ID NO: 32 QVQLQESGPGLVKPSQTLSLTCTVSGFSLS RYSVH WVRQPPGKGLEWIG M IWDGGNTDYAAPVKG RFTISRDDSKNTLYLQMNSLKTEDTAVYYCAR NPL YFSYAMDY WGQGTLVTVSS .
  • the HCDR2 sequence obtained after mutation is as follows:
  • the heavy chain variable regions hu2G3 VH1.1 and hu2G3 VH1.2 were combined with the light chain variable region hu2G3 VL to form new optimized humanized antibodies, see Table 7.
  • the humanized antibodies described above were tested for affinity (refer to the capture ELISA in Example 2), and the test results showed that the humanized molecules can bind to OX4O.
  • the light chain variable region sequence as described above was combined with the light chain constant region sequence to form the final light chain sequences, and the heavy chain variable region sequence as described above was combined with the heavy chain constant region to form the final heavy chain sequences.
  • the specific light and heavy chain constant regions are not limited to the antibody constant regions disclosed in the present disclosure. Other light and heavy chain constant regions and the mutants thereof known in the art can also be used to increase the performance of the antibody.
  • the exemplary constant regions are as follows:
  • IgG1 heavy chain constant region SEQ ID NO: 35 ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGV HTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEP KSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVS HEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGK EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTC LVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSPGK; kappa light chain constant region: SEQ ID NO: 36 RTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWK
  • amino acid sequences of exemplary full-length 2G3 and hu4B5-V7 (also known as 4B5) antibody are as follows:
  • 2G3 heavy chain SEQ ID NO: 37 QVQLQESGPGLVKPSETLSLTCTVSGYSISRYSVHWVRQPPGKGLEWIGM IWDGGNTDYNAALKSRFTISRDNAKNTLYLQMNSLRAEDTAVYYCARNPL YFSYAMDYWGQGTLVTVSS ASTKGPSVFPLAPSSKSTSGGTAALGCLVKD YFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTY ICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPK DTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL DSDGS
  • the positive control GPX4 was prepared with reference to 1A7. gr.1 taught in the patent WO2015153513, and the heavy and light chain amino acid sequences thereof are as follows:
  • GPX4 heavy chain SEQ ID NO: 41 EVQLVQSGAEVKKPGASVKVSCKASGYTFTDSYMSWVRQAPGQGLEWIGD MYPDNGDSSYNQKFRERVTITRDTSTSTAYLELSSLRSEDTAVYYCVLAP RWYFSVWGQGTLVTVSS ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYF PEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYIC NVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDT LMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTY RVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYT LPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDS DGS
  • the Biacore method is a well-recognized method for objectively detecting the affinity and kinetics between proteins.
  • the recombinant anti-OX40 antibody to be tested of the present invention was covalently connected to the CM5 (GE) chip by NHS standard amino coupling method.
  • a series of concentration gradients of human OX40-His protein (sinobiological # 10481-H08H) diluted in the same buffer were then loaded into each cycle at a flow rate of 30 ⁇ L/min.
  • the chip was regenerated using regeneration reagent provided in the kit.
  • the antigen-antibody binding kinetics was traced for 3 minutes, and the dissociation kinetics was traced for 10 minutes.
  • the resulting data were analyzed by using BIAevaluation software (GE) with 1:1 (Langmuir) binding model.
  • the ka (kon), kd (koff) and KD values of the chimeric antibodies determined by this method are shown in the table below.
  • the well-plate was coated with the OX40 ligand (Acrobiosystem, OXL-H52Q8), blocked, added the gradient-diluted antibody to be tested (the antibody was diluted in a solution comprising human Bio-OX40-FC (Acrobiosystem, OX40-H5255, labeled with biotin), pre-incubated for 40 minutes, and then added into the plate), incubated for 40 minute, and washed.
  • SA-HRP Jackson Immunoresearch, 016-030-084
  • the developing solution and stop solution were added, and the OD value was measured. The results are shown in the table below.
  • the well-plate was coated with anti-CD3 antibody (Chempartner, A05-001), placed at 4° C. overnight and washed for 3 times with PBS; Jurkat-NF-Kb luc-hOX40 cells (ATCC, TIB-152, a stable cell line constructed by Shanghai ChemPartner) and Raji cells (ATCC, CCL-86) were harvested. The two kinds of cells were re-suspended and mixed. 50 ⁇ l/well of the diluted antibody to be tested and 50 ⁇ l/well of the mixed two kinds of cells were added to the plate, and the plate was incubated at 37° C. in 5% CO 2 incubator for 5 hours.
  • One-Glo TM luciferase reagent (Promega, Cat# E6120) was added to each well, and incubated at room temperature for more than 3 minutes. The luminescent signal was measured on the instrument, RLU reading was recorded. The results are shown in Table 12. The results show that both 2G3 and 4B5 can effectively activate the reporter gene.
  • CD4+ memory T cells were isolated and added together with the antibody to be tested to a 96-well plate coated with anti-CD3 antibody (Chempartner, A05-001), co-incubated at 37° C. for 72 hours, the supernatant was collected to detect IFN- ⁇ .
  • the results are shown in FIG. 2 and Table 13. The results show that GPX4, 4B5 and 2G3 can significantly enhance the release of IFN- ⁇ , and 2G3 can achieve the maximum stimulating effect at lOng/mL.
  • B-hTNFRSF4 (OX40) humanized mice (B-hTNFRSF4 (OX40) humanized mice, from Biocytogen Jiangsu Gene Biotechnology Co., Ltd.), female, 17 to 20 g, 6 to 7 week old.
  • the MC38 tumor cells 7 at logarithmic growth phase were collected (purchased from Nanjing Yinhe Biomedicine Co., Ltd.), the cell concentration was adjusted to 5 ⁇ 10 6 /mL with PBS buffer, and 0.1 mL of cell suspension was inoculated to the flanks of OX40 mice. The mice were monitored after inoculation for the growth of tumors. On day 7 after inoculation, the average tumor volume in the flank of tumor-bearing mice reached 102.5 mm 3 . Mice were divided into groups according to the tumor size, administrated and monitored. The grouping information is shown in Table 14.
  • the OX40 humanized antibody was tested for its inhibitory effect on the growth of MC38 colon cancer cell xenograft in mice.
  • tumor was measured twice a week using a caliper.
  • Tumor growth xenograft TGI (%) [1 ⁇ T/C] ⁇ 100.
  • the weight of all tumor-bearing mice was measured twice a week.
  • the average tumor volume of IgG1 control group reached 1732.593 mm 3 ; and the average tumor volume of the test compound 2G3 low-dose administration group (0.3 mg/kg), medium-dose administration group (2G3, lmg/kg) and high-dose administration group (2G, 3 mg/kg) reached 930.37 mm 3 , 303.49 mm 3 and 155.79 mm 3 , respectively.
  • the medium and high-dose group significantly inhibited the tumor growth, when compared with the control group (**P ⁇ 0.01), and a preliminary dose-dependent relationship was observed, with the tumor growth inhibition rate of up to 49%, 88% and 97.0%, respectively.
  • the average tumor volume in the 3 mg/kg GPX4 group was 362.47 mm 3 , which was significantly different from that of control group, and 3 mg/kg GPX4 group also showed significant tumor growth inhibition (*P ⁇ 0.05), with the tumor growth inhibition rate of up to 84%.
  • mice were euthanized, and the subcutaneous xenograft mass was removed from the tumor-bearing mice and weighed.
  • the average weight of tumor mass in the control group was 1.568 g; as for the test compound 2G3 in the low-dose group (0.3 mg/kg), medium-dose group (lmg/kg) and high-dose group (3 mg/kg), the average tumor weight reached 0.926 g, 0.251 g and 0.181 g, respectively; the medium and high-dose group significantly inhibited the tumor growth, when compared with the control group (**P ⁇ 0.01).
  • the average tumor weight was 0.372 g, which was significantly different from that of the control group, and GPX4 (3 mg/kg) administration group also showed a significant inhibitory effect on the growth of MC38 tumor cells (**P ⁇ 0.01).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Oncology (AREA)
  • Plant Pathology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Mycology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US17/278,259 2018-09-26 2019-09-25 An anti-ox40 antibody, antigen-binding fragment thereof, and the pharmaceutical use Pending US20210355229A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN201811128669.3 2018-09-26
CN201811128669 2018-09-26
CN201811417666 2018-11-26
CN201811417666.1 2018-11-26
PCT/CN2019/107787 WO2020063660A1 (zh) 2018-09-26 2019-09-25 抗ox40抗体、其抗原结合片段及其医药用途

Publications (1)

Publication Number Publication Date
US20210355229A1 true US20210355229A1 (en) 2021-11-18

Family

ID=69952440

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/278,259 Pending US20210355229A1 (en) 2018-09-26 2019-09-25 An anti-ox40 antibody, antigen-binding fragment thereof, and the pharmaceutical use

Country Status (11)

Country Link
US (1) US20210355229A1 (pt)
EP (1) EP3858857A1 (pt)
JP (1) JP2022502417A (pt)
KR (1) KR20210069058A (pt)
CN (1) CN112513088B (pt)
AU (1) AU2019347934A1 (pt)
BR (1) BR112021005169A2 (pt)
CA (1) CA3113541A1 (pt)
MX (1) MX2021003168A (pt)
TW (1) TW202035455A (pt)
WO (1) WO2020063660A1 (pt)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114106173A (zh) * 2020-08-26 2022-03-01 上海泰槿生物技术有限公司 抗ox40抗体、其药物组合物及应用
CN114106174A (zh) * 2020-08-27 2022-03-01 苏州景涞医疗科技有限公司 低毒性抗ox40抗体、其药物组合物及应用
CN114515335A (zh) * 2020-11-19 2022-05-20 百奥泰生物制药股份有限公司 抗ox40抗体在治疗肿瘤或癌症中的应用

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
IL85035A0 (en) 1987-01-08 1988-06-30 Int Genetic Eng Polynucleotide molecule,a chimeric antibody with specificity for human b cell surface antigen,a process for the preparation and methods utilizing the same
US6407213B1 (en) 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
ES2532910T3 (es) 1998-04-02 2015-04-01 Genentech, Inc. Variantes de anticuerpos y fragmentos de los mismos
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
CN101668776A (zh) * 2007-02-27 2010-03-10 健泰科生物技术公司 拮抗剂ox40抗体及其在炎性和自身免疫疾病的治疗中的用途
CN103221427B (zh) * 2010-08-23 2016-08-24 德克萨斯州立大学董事会 抗ox40抗体和使用其的方法
EA036047B1 (ru) * 2011-07-11 2020-09-18 Икнос Сайенсиз Са Антитела, которые связываются с ox40, и их применение
GB201116092D0 (en) 2011-09-16 2011-11-02 Bioceros B V Antibodies and uses thereof
MA39355B2 (fr) * 2014-03-31 2021-04-30 Genentech Inc Anticorps anti-ox40 et procédés d'utilisation correspondants
LT3126394T (lt) 2014-03-31 2020-01-27 F. Hoffmann-La Roche Ag Antikūnai prieš ox40 ir jų naudojimo būdai
EP3292152A1 (en) * 2015-05-07 2018-03-14 Agenus Inc. Anti-ox40 antibodies and methods of use thereof
PE20180926A1 (es) * 2015-05-29 2018-06-08 Bristol Myers Squibb Co Anticuerpos contra el miembro 4 de la superfamilia del receptor del factor de necrosis tumoral (ox40) y sus usos
AU2016364891A1 (en) 2015-12-03 2018-06-07 Agenus Inc. Anti-OX40 antibodies and methods of use thereof
US10442866B1 (en) 2019-01-23 2019-10-15 Beijing Mabworks Biotech Co. Ltd Antibodies binding OX40 and uses thereof

Also Published As

Publication number Publication date
AU2019347934A1 (en) 2021-04-08
CN112513088B (zh) 2023-05-16
WO2020063660A1 (zh) 2020-04-02
CN112513088A (zh) 2021-03-16
BR112021005169A2 (pt) 2021-06-15
KR20210069058A (ko) 2021-06-10
EP3858857A1 (en) 2021-08-04
CA3113541A1 (en) 2020-04-02
TW202035455A (zh) 2020-10-01
MX2021003168A (es) 2021-05-14
JP2022502417A (ja) 2022-01-11

Similar Documents

Publication Publication Date Title
US11981733B2 (en) LAG-3 antibody, antigen-binding fragment thereof, and pharmaceutical application thereof
US11365255B2 (en) PD-1 antibody, antigen-binding fragment thereof, and medical application thereof
EP3712170A1 (en) Cd96 antibody, antigen-binding fragment and pharmaceutical use thereof
JP2021513331A (ja) 抗b7−h4抗体、その抗原結合断片及びその医薬用途
EP3741777A1 (en) Pd-l1 antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
EP3632932A1 (en) Anti-cd40 antibody, antigen binding fragment thereof and medical use thereof
EP3744734A1 (en) Anti-4-1bb antibody, antigen-binding fragment thereof and medical use thereof
US20210355229A1 (en) An anti-ox40 antibody, antigen-binding fragment thereof, and the pharmaceutical use
CN110790839A (zh) 抗pd-1抗体、其抗原结合片段及医药用途
TW201916890A (zh) 抗pd-1抗體和抗lag-3抗體聯合在製備治療腫瘤的藥物中的用途
WO2021190582A1 (zh) 一种抗ox40抗体药物组合物及其用途

Legal Events

Date Code Title Description
AS Assignment

Owner name: JIANGSU HENGRUI MEDICINE CO., LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIAO, CHENG;XU, ZUPENG;JIANG, JIAHUA;AND OTHERS;REEL/FRAME:055681/0062

Effective date: 20210317

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED